1
|
Liang S, Hu Z. Unveiling the predictive power of biomarkers in traumatic brain injury: A narrative review focused on clinical outcomes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024. [PMID: 39687991 DOI: 10.5507/bp.2024.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Traumatic brain injury (TBI) has long-term consequences, including neurodegenerative disease risk. Current diagnostic tools are limited in detecting subtle brain damage. This review explores emerging biomarkers for TBI, including those related to neuronal injury, inflammation, EVs, and ncRNAs, evaluating their potential to predict clinical outcomes like mortality, recovery, and cognitive impairment. It addresses challenges and opportunities for implementing biomarkers in clinical practice, aiming to improve TBI diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Sitao Liang
- Neurosurgery Department, Zhongshan City People's Hospital, Zhongshan, 528400, China
| | - Zihui Hu
- Neurosurgery Department, Zhongshan City People's Hospital, Zhongshan, 528400, China
| |
Collapse
|
2
|
Yue Y, Deng B, Zeng Y, Li W, Qiu X, Hu P, Shen L, Ruan T, Zhou R, Li S, Ying J, Xiong T, Qu Y, Luan Z, Mu D. Oligodendrocyte Progenitor Cell Transplantation Reduces White Matter Injury in a Fetal Goat Model. CNS Neurosci Ther 2024; 30:e70178. [PMID: 39690788 DOI: 10.1111/cns.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Preterm white matter injury (PWMI) is the most common type of brain injury in preterm infants, in which, oligodendrocyte progenitor cells (OPCs) are predominantly damaged. In this study, human OPCs (hOPCs) were administered to a fetal goat model of PWMI to examine the differentiation potential and therapeutic effects of the cells on PWMI. METHODS Preterm goat fetuses were subjected to hypoxic-ischemia (HI) via intermittent umbilical cord occlusion (5 min × 5). Twenty million hOPCs were administered via a nasal catheter 12 h after an HI insult, and brain tissues were collected 14 or 21 days after the HI insult. Myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) were detected by immunofluorescence and western blotting techniques. The percentage of myelinated nerve fibers and g-ratio were examined using transmission electron microscopy. Inflammatory cells were detected by immunohistochemistry. Inflammatory and neurotrophic factors were measured using enzyme-linked immunosorbent assay. RESULTS Our results showed that intermittent umbilical cord occlusion induced PWMI in fetal goats. Transplanted hOPCs can survive in periventricular and subcortical white matter. Further, transplanted hOPCs expressed markers of mature oligodendrocytes (MBP and MAG) and few cells expressed markers of preoligodendrocytes (NG2 and A2B5), suggesting that these cells can differentiate into mature oligodendrocytes in the brain. In addition, hOPCs administration increased MBP and MAG levels, percentage of myelinated nerve fibers, and thickness of the myelin sheath, indicating a reduction in PWMI. Furthermore, hOPCs did not increase the inflammatory response after HI. Interestingly, hOPC administration decreased tumor necrosis factor-alpha and increased glial-derived neurotrophic factor and brain-derived neurotrophic factor levels after HI, suggesting that additional mechanisms mediate the inflammatory microenvironment and neuroprotective effects. CONCLUSIONS Exogenous hOPCs can differentiate into mature oligodendrocytes in fetal goats and alleviate HI-induced PWMI. Transplantation of hOPCs is a promising strategy for treating PWMI.
Collapse
Affiliation(s)
- Yan Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Bixin Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Yan Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Wenxing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Xia Qiu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Peng Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - LiuHong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tiechao Ruan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Ruixi Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Shiping Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Junjie Ying
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Tao Xiong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Yi Qu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Zuo Luan
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dezhi Mu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Kråkenes T, Sandvik CE, Ytterdal M, Gavasso S, Evjenth EC, Bø L, Kvistad CE. The Therapeutic Potential of Exosomes from Mesenchymal Stem Cells in Multiple Sclerosis. Int J Mol Sci 2024; 25:10292. [PMID: 39408622 PMCID: PMC11477223 DOI: 10.3390/ijms251910292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Although treatment for multiple sclerosis (MS) has undergone a revolution in the last decades, at least two important barriers remain: alleviation of innate inflammation driving disease progression and promotion of remyelination and neural regeneration. Mesenchymal stem cells (MSCs) possess immunomodulatory properties and promote remyelination in murine MS models. The main therapeutic mechanism has, however, been attributed to their potent paracrine capacity, and not to in vivo tissue implantation. Studies have demonstrated that exosomes released as part of the cells' secretome effectively encapsulate the beneficial properties of MSCs. These membrane-enclosed nanoparticles contain a variety of proteins and nucleic acids and serve as mediators of intercellular communication. In vitro studies have demonstrated that exosomes from MSCs modulate activated microglia from an inflammatory to an anti-inflammatory phenotype and thereby dampen the innate inflammation. Rodent studies have also demonstrated potent immunomodulation and remyelination with improved outcomes following exosome administration. Thus, exosomes from MSCs may represent a potential cell-free treatment modality to prevent disease progression and promote remyelination in MS. In this narrative review, we summarize the current knowledge of exosomes from MSCs as a potential treatment for MS and discuss the remaining issues before successful translation into clinical trials.
Collapse
Affiliation(s)
- Torbjørn Kråkenes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Casper Eugen Sandvik
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Marie Ytterdal
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Sonia Gavasso
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Elisabeth Claire Evjenth
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Lars Bø
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Christopher Elnan Kvistad
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| |
Collapse
|
4
|
Fang X, Zhou D, Wang X, Ma Y, Zhong G, Jing S, Huang S, Wang Q. Exosomes: A Cellular Communication Medium That Has Multiple Effects On Brain Diseases. Mol Neurobiol 2024; 61:6864-6892. [PMID: 38356095 DOI: 10.1007/s12035-024-03957-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Exosomes, as membranous vesicles generated by multiple cell types and secreted to extracellular space, play a crucial role in a range of brain injury-related brain disorders by transporting diverse proteins, RNA, DNA fragments, and other functional substances. The nervous system's pathogenic mechanisms are complicated, involving pathological processes like as inflammation, apoptosis, oxidative stress, and autophagy, all of which result in blood-brain barrier damage, cognitive impairment, and even loss of normal motor function. Exosomes have been linked to the incidence and progression of brain disorders in recent research. As a result, a thorough knowledge of the interaction between exosomes and brain diseases may lead to the development of more effective therapeutic techniques that may be implemented in the clinic. The potential role of exosomes in brain diseases and the crosstalk between exosomes and other pathogenic processes were discussed in this paper. Simultaneously, we noted the delicate events in which exosomes as a media allow the brain to communicate with other tissues and organs in physiology and disease, and compiled a list of natural compounds that modulate exosomes, in order to further improve our understanding of exosomes and propose new ideas for treating brain disorders.
Collapse
Affiliation(s)
- Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Dishu Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510405, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 510405, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shangwen Jing
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shuiqing Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| |
Collapse
|
5
|
Ramos Ferrer P, Sakiyama-Elbert S. Affinity-based drug delivery systems for the central nervous system: exploiting molecular interactions for local, precise targeting. J Neural Eng 2024; 21:041004. [PMID: 39059438 DOI: 10.1088/1741-2552/ad680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/26/2024] [Indexed: 07/28/2024]
Abstract
Objective: The effective treatment of central nervous system (CNS) disorders remains a significant challenge, primarily due to its molecular and structural complexity. Clinical translation of promising therapeutic agents is limited by the lack of optimal drug delivery systems capable of targeted, localized release of drugs to the brain and spinal cord.Approach: This review provides an overview of the potential of affinity-based drug delivery systems, which leverage molecular interactions to enhance the delivery and efficacy of therapeutic agents within the CNS.Main results: Various approaches, including hydrogels, micro- and nanoparticles, and functionalized biomaterials, are examined for their ability to provide local, sustained release of proteins, growth factors and other drugs.Significance: Furthermore, we present a detailed analysis of design considerations for developing effective affinity-based systems, incorporating insights from both existing literature and our group's research. These considerations include the biochemical modification of delivery vehicles and the optimization of physical and chemical properties to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Pablo Ramos Ferrer
- Department of Chemical Engineering, University of Washington, Seattle, WA, United States of America
| | - Shelly Sakiyama-Elbert
- Department of Chemical Engineering, University of Washington, Seattle, WA, United States of America
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
6
|
Gotoh S, Kawabori M, Fujimura M. Intranasal administration of stem cell-derived exosomes for central nervous system diseases. Neural Regen Res 2024; 19:1249-1255. [PMID: 37905871 PMCID: PMC11467946 DOI: 10.4103/1673-5374.385875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Exosomes, lipid bilayer-enclosed small cellular vesicles, are actively secreted by various cells and play crucial roles in intercellular communication. These nanosized vesicles transport internalized proteins, mRNA, miRNA, and other bioactive molecules. Recent findings have provided compelling evidence that exosomes derived from stem cells hold great promise as a therapeutic modality for central nervous system disorders. These exosomes exhibit multifaceted properties including anti-apoptotic, anti-inflammatory, neurogenic, and vasculogenic effects. Furthermore, exosomes offer several advantages over stem cell therapy, such as high preservation capacity, low immunogenicity, the ability to traverse the blood-brain barrier, and the potential for drug encapsulation. Consequently, researchers have turned their attention to exosomes as a novel therapeutic avenue. Nonetheless, akin to the limitations of stem cell treatment, the limited accumulation of exosomes in the injured brain poses a challenge to their clinical application. To overcome this hurdle, intranasal administration has emerged as a non-invasive and efficacious route for delivering drugs to the central nervous system. By exploiting the olfactory and trigeminal nerve axons, this approach enables the direct transport of therapeutics to the brain while bypassing the blood-brain barrier. Notably, exosomes, owing to their small size, can readily access the nerve pathways using this method. As a result, intranasal administration has gained increasing recognition as an optimal therapeutic strategy for exosome-based treatments. In this comprehensive review, we aim to provide an overview of both basic and clinical research studies investigating the intranasal administration of exosomes for the treatment of central nervous system diseases. Furthermore, we elucidate the underlying therapeutic mechanisms and offer insights into the prospect of this approach.
Collapse
Affiliation(s)
- Shuho Gotoh
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| |
Collapse
|
7
|
Shamshiripour P, Rahnama M, Nikoobakht M, Rad VF, Moradi AR, Ahmadvand D. Extracellular vesicles derived from dendritic cells loaded with VEGF-A siRNA and doxorubicin reduce glioma angiogenesis in vitro. J Control Release 2024; 369:128-145. [PMID: 38522817 DOI: 10.1016/j.jconrel.2024.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Numerous attempts have been devoted to designing anti-angiogenic agents as a strategy to slow tumor growth and progression. Clinical applications of conventional anti-angiogenic agents face some challenges, e.g., off-target effects for TKIs and also low solid tumor penetration for mAbs. Furthermore, although anti-angiogenic therapy provides a normalization window for better chemo-RT response, in long-term treatments, tumor hypoxia as a result of total removal of VEGF-A by mAbs from the TME or complete blockade of TK receptors induces over-activation of compensatory angiogenic pathways, causing escape. Herein, we investigate the efficacy of si-DOX-DC-EVs to reduce glioma angiogenesis and invasiveness. METHODS Mature DCs were generated from PBMC and EVs were isolated from the DCs culture media. siRNA and Doxorubicin were loaded into EVs by EP and incubation. Afterward, the uptake of DC-EVs was assessed by flow cytometry, and the subcellular localization of EVs was tested by confocal imaging. Tube formation assay was performed to assess the efficacy of si-DOX-DC-EVs to reduce tumor angiogenesis which was analyzed by DHM. Morphometric analysis of apoptotic cells was performed by DHM and confocal imaging and further, ELISA was performed for hypoxia-related and angiogenic cytokines. The impact of our theranostic system "si-DOX-DC-MVs" on the formation of vascular mimics, colonies, and invasion of C6 cells was checked in vitro. Afterward, orthotropic rat models of glioma were generated and the optimal administration route was selected by in vivo fluorescent analysis. Then, the microvessel density, vimentin expression, and accumulation of immune cells in tumoral tissues were assessed by IHC. Finally, necropsy and autopsy analyses were performed to check the safety of our theranostic agent. RESULTS DC-EVs loaded with si-DOX-DC-EVs were successfully uptaken by cells with different subcellular trafficking for MVs and exosomes, reduced tumor angiogenesis in DHM analysis, and induced apoptosis in tumoral cells. Moreover, using DHM, we performed a detailed label-free analysis of tip cells which suggested that the tip cells in si-DC-MV treatments lost their geometrical migration capacity to form tube-like structures. Furthermore, the ELISAs performed highlighted that there is a mild overactivation of compensatory Tie2/Ang2 pathway after VEGF-A blockade which confers with severe hypoxia and sustains normal angiogenesis which is the optimal goal of anti-angiogenesis therapy for cancer to avoid resistance.The results of our VM analyses indicated that si-DOX-DC-MVs completely inhibited VM process. Moreover, the invasion, migration, and colony formation of the C6 cells treated with si-DOX-MVs were the least among all treatments. IN was the optimal route of administration. The MVD analyses indicated that si-DOX-DC-MVs reduced the number of tumoral microvessels and normalized vessel morphology. Intense CD8+ T cells were observed near the tumoral vessels in the si-DOX-DC-MVs group and with minimal activation of MT (low Vimentin expression). Necropsy and toxicology results proved that the theranostic system proposed is safe. CONCLUSIONS DC-EVs loaded with VEGF-A siRNA and Doxorubicin were more potent than BV alone as a multi-disciplinary strategy that combats glioma growth by cytotoxic impacts of DOX and inhibits angiogenesis by VEGF-A siRNAs with excess immunologic benefits from DC-EVs. This next-generation anti-angiogenic agent normalizes tumor vessel density rather than extensively eliminating tumor vessels causing hypoxia and mesenchymal transition.
Collapse
Affiliation(s)
- Parisa Shamshiripour
- Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Molecular Imaging Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mehrana Rahnama
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Mehdi Nikoobakht
- Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Neurosurgery, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Vahideh Farzam Rad
- Department of Physics, Institute for Advanced Studies in Basic Sciences, (IASBS), Zanjan, Iran
| | - Ali-Reza Moradi
- Department of Physics, Institute for Advanced Studies in Basic Sciences, (IASBS), Zanjan, Iran; School of NanoScience, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Davoud Ahmadvand
- Department of Molecular Imaging Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
8
|
Fallahi S, Zangbar HS, Farajdokht F, Rahbarghazi R, Mohaddes G, Ghiasi F. Exosomes as a therapeutic tool to promote neurorestoration and cognitive function in neurological conditions: Achieve two ends with a single effort. CNS Neurosci Ther 2024; 30:e14752. [PMID: 38775149 PMCID: PMC11110007 DOI: 10.1111/cns.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/16/2024] [Accepted: 04/13/2024] [Indexed: 05/25/2024] Open
Abstract
Exosomes possess a significant role in intercellular communications. In the nervous system, various neural cells release exosomes that not only own a role in intercellular communications but also eliminate the waste of cells, maintain the myelin sheath, facilitate neurogenesis, and specifically assist in normal cognitive function. In neurological conditions including Parkinson's disease (PD), Alzheimer's disease (AD), traumatic brain injury (TBI), and stroke, exosomal cargo like miRNAs take part in the sequela of conditions and serve as a diagnostic tool of neurological disorders, too. Exosomes are not only a diagnostic tool but also their inhibition or administration from various sources like mesenchymal stem cells and serum, which have shown a worthy potential to treat multiple neurological disorders. In addition to neurodegenerative manifestations, cognitive deficiencies are an integral part of neurological diseases, and applying exosomes in improving both aspects of these diseases has been promising. This review discusses the status of exosome therapy in improving neurorestorative and cognitive function following neurological disease.
Collapse
Affiliation(s)
- Solmaz Fallahi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fereshteh Farajdokht
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Gisou Mohaddes
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Biomedical EducationCalifornia Health Sciences University, College of Osteopathic MedicineClovisCaliforniaUSA
| | - Fariba Ghiasi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
9
|
Ding S, Kim YJ, Huang KY, Um D, Jung Y, Kong H. Delivery-mediated exosomal therapeutics in ischemia-reperfusion injury: advances, mechanisms, and future directions. NANO CONVERGENCE 2024; 11:18. [PMID: 38689075 PMCID: PMC11061094 DOI: 10.1186/s40580-024-00423-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Ischemia-reperfusion injury (IRI) poses significant challenges across various organ systems, including the heart, brain, and kidneys. Exosomes have shown great potentials and applications in mitigating IRI-induced cell and tissue damage through modulating inflammatory responses, enhancing angiogenesis, and promoting tissue repair. Despite these advances, a more systematic understanding of exosomes from different sources and their biotransport is critical for optimizing therapeutic efficacy and accelerating the clinical adoption of exosomes for IRI therapies. Therefore, this review article overviews the administration routes of exosomes from different sources, such as mesenchymal stem cells and other somatic cells, in the context of IRI treatment. Furthermore, this article covers how the delivered exosomes modulate molecular pathways of recipient cells, aiding in the prevention of cell death and the promotions of regeneration in IRI models. In the end, this article discusses the ongoing research efforts and propose future research directions of exosome-based therapies.
Collapse
Affiliation(s)
- Shengzhe Ding
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Yu-Jin Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Kai-Yu Huang
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - Daniel Um
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Department of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjoon Kong
- Chemical & Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA.
- Bioengineering, University of Illinois, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, 61801, USA.
- Chan Zuckerberg Biohub-Chicago, Chicago, USA.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
10
|
Zhang L, Bai W, Peng Y, Lin Y, Tian M. Human umbilical cord mesenchymal stem cell-derived exosomes provide neuroprotection in traumatic brain injury through the lncRNA TUBB6/Nrf2 pathway. Brain Res 2024; 1824:148689. [PMID: 38030103 DOI: 10.1016/j.brainres.2023.148689] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/16/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023]
Abstract
Recently, human umbilical cord mesenchymal stem cell (HucMSC) is a new focus of research in neurological diseases, and the beneficial effect of HucMSC is mediated by paracrine factors which are transported by exosome. Our previous study has shown that HucMSC-derived exosome could provide neuroprotection after traumatic brain injury (TBI). However, the underlying mechanisms were not fully understood. In the present study, we found that administration of exosome suppressed TBI-induced inflammation and ferroptosis. In addition, exosome activated the long non-coding ribonucleic acid (lncRNA) TUBB6/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway after TBI. However, exosome partly failed to provide neuroprotection following TBI when TUBB6 was knockdown. Importantly, exosome treatment also decreased neuron cell death, suppressed inflammation, inhibited ferroptosis and activated the lncRNA TUBB6/Nrf2 pathway after TBI in vitro. Taken together, our results provided the first evidence that HucMSC-derived exosome played a key role in neuroprotection after TBI through the lncRNA TUBB6/Nrf2 pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Wanshan Bai
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yaonan Peng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yixing Lin
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Mi Tian
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, PR China.
| |
Collapse
|
11
|
Li S, Qiu N, Ni A, Hamblin MH, Yin KJ. Role of regulatory non-coding RNAs in traumatic brain injury. Neurochem Int 2024; 172:105643. [PMID: 38007071 PMCID: PMC10872636 DOI: 10.1016/j.neuint.2023.105643] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/19/2023] [Indexed: 11/27/2023]
Abstract
Traumatic brain injury (TBI) is a potentially fatal health event that cannot be predicted in advance. After TBI occurs, it can have enduring consequences within both familial and social spheres. Yet, despite extensive efforts to improve medical interventions and tailor healthcare services, TBI still remains a major contributor to global disability and mortality rates. The prompt and accurate diagnosis of TBI in clinical contexts, coupled with the implementation of effective therapeutic strategies, remains an arduous challenge. However, a deeper understanding of changes in gene expression and the underlying molecular regulatory processes may alleviate this pressing issue. In recent years, the study of regulatory non-coding RNAs (ncRNAs), a diverse class of RNA molecules with regulatory functions, has been a potential game changer in TBI research. Notably, the identification of microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and other ncRNAs has revealed their potential as novel diagnostic biomarkers and therapeutic targets for TBI, owing to their ability to regulate the expression of numerous genes. In this review, we seek to provide a comprehensive overview of the functions of regulatory ncRNAs in TBI. We also summarize regulatory ncRNAs used for treatment in animal models, as well as miRNAs, lncRNAs, and circRNAs that served as biomarkers for TBI diagnosis and prognosis. Finally, we discuss future challenges and prospects in diagnosing and treating TBI patients in the clinical settings.
Collapse
Affiliation(s)
- Shun Li
- Department of Neurology, School of Medicine, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA
| | - Na Qiu
- Department of Neurology, School of Medicine, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA
| | - Andrew Ni
- Warren Alpert Medical School, Brown University, 222 Richmond Street, Providence, RI, 02903, USA
| | - Milton H Hamblin
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, 1212 Webber Hall, 900 University Avenue, Riverside, CA, 92521, USA
| | - Ke-Jie Yin
- Department of Neurology, School of Medicine, University of Pittsburgh, S514 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
12
|
Wu H, Fan Y, Zhang M. Advanced Progress in the Role of Adipose-Derived Mesenchymal Stromal/Stem Cells in the Application of Central Nervous System Disorders. Pharmaceutics 2023; 15:2637. [PMID: 38004615 PMCID: PMC10674952 DOI: 10.3390/pharmaceutics15112637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Currently, adipose-derived mesenchymal stromal/stem cells (ADMSCs) are recognized as a highly promising material for stem cell therapy due to their accessibility and safety. Given the frequently irreversible damage to neural cells associated with CNS disorders, ADMSC-related therapy, which primarily encompasses ADMSC transplantation and injection with exosomes derived from ADMSCs or secretome, has the capability to inhibit inflammatory response and neuronal apoptosis, promote neural regeneration, as well as modulate immune responses, holding potential as a comprehensive approach to treat CNS disorders and improve prognosis. Empirical evidence from both experiments and clinical trials convincingly demonstrates the satisfactory safety and efficacy of ADMSC-related therapies. This review provides a systematic summary of the role of ADMSCs in the treatment of central nervous system (CNS) disorders and explores their therapeutic potential for clinical application. ADMSC-related therapy offers a promising avenue to mitigate damage and enhance neurological function in central nervous system (CNS) disorders. However, further research is necessary to establish the safety and efficacy of clinical ADMSC-based therapy, optimize targeting accuracy, and refine delivery approaches for practical applications.
Collapse
Affiliation(s)
- Haiyue Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; (H.W.); (Y.F.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
13
|
Mehryab F, Taghizadeh F, Goshtasbi N, Merati F, Rabbani S, Haeri A. Exosomes as cutting-edge therapeutics in various biomedical applications: An update on engineering, delivery, and preclinical studies. Biochimie 2023; 213:139-167. [PMID: 37207937 DOI: 10.1016/j.biochi.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/29/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Exosomes are cell-derived nanovesicles, circulating in different body fluids, and acting as an intercellular mechanism. They can be purified from culture media of different cell types and carry an enriched content of various protein and nucleic acid molecules originating from their parental cells. It was indicated that the exosomal cargo can mediate immune responses via many signaling pathways. Over recent years, the therapeutic effects of various exosome types were broadly investigated in many preclinical studies. Herein, we present an update on recent preclinical studies on exosomes as therapeutic and/or delivery agents for various applications. The exosome origin, structural modifications, natural or loaded active ingredients, size, and research outcomes were summarized for various diseases. Overall, the present article provides an overview of the latest exosome research interests and developments to clear the way for the clinical study design and application.
Collapse
Affiliation(s)
- Fatemeh Mehryab
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Taghizadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Merati
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Yang ZL, Liang ZY, Lin YK, Lin FB, Rao J, Xu XJ, Wang CH, Chen CM. Efficacy of extracellular vesicles of different cell origins in traumatic brain injury: A systematic review and network meta-analysis. Front Neurosci 2023; 17:1147194. [PMID: 37065922 PMCID: PMC10090410 DOI: 10.3389/fnins.2023.1147194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundThere was still no effective treatment for traumatic brain injury (TBI). Recently, many preclinical studies had shown promising efficacy of extracellular vesicles (EVs) from various cell sources. Our aim was to compare which cell-derived EVs were most effective in treating TBI through a network meta-analysis.MethodsWe searched four databases and screened various cell-derived EVs for use in preclinical studies of TBI treatment. A systematic review and network meta-analysis were conducted for two outcome indicators, modified Neurological Severity Score (mNSS) and Morris Water Maze (MWM), and they were ranked by the surface under the cumulative ranking curves (SUCRA). Bias risk assessment was performed with SYRCLE. R software (version 4.1.3, Boston, MA, USA) was used for data analysis.ResultsA total of 20 studies were included in this study, involving 383 animals. Astrocyte-derived extracellular vesicles (AEVs) ranked first in response to mNSS at day 1 (SUCRA: 0.26%), day 3 (SUCRA: 16.32%), and day 7 (SUCRA: 9.64%) post-TBI. Extracellular vesicles derived from mesenchymal stem cells (MSCEVs) were most effective in mNSS assessment on day 14 (SUCRA: 21.94%) and day 28 (SUCRA: 6.26%), as well as MWM’s escape latency (SUCRA: 6.16%) and time spent in the target quadrant (SUCRA: 86.52%). The result of mNSS analysis on day 21 showed that neural stem cell-derived extracellular vesicles (NSCEVs) had the best curative effect (SUCRA: 6.76%).ConclusionAEVs may be the best choice to improve early mNSS recovery after TBI. The efficacy of MSCEVs may be the best in the late mNSS and MWM after TBI.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/, identifier CRD42023377350.
Collapse
|
15
|
Kodali M, Madhu LN, Reger RL, Milutinovic B, Upadhya R, Gonzalez JJ, Attaluri S, Shuai B, Gitai DLG, Rao S, Choi JM, Jung SY, Shetty AK. Intranasally administered human MSC-derived extracellular vesicles inhibit NLRP3-p38/MAPK signaling after TBI and prevent chronic brain dysfunction. Brain Behav Immun 2023; 108:118-134. [PMID: 36427808 PMCID: PMC9974012 DOI: 10.1016/j.bbi.2022.11.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/21/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Traumatic brain injury (TBI) leads to lasting brain dysfunction with chronic neuroinflammation typified by nucleotide-binding domain leucine-rich repeat and pyrin domain-containing receptor 3 (NLRP3) inflammasome activation in microglia. This study probed whether a single intranasal (IN) administration of human mesenchymal stem cell-derived extracellular vesicles (hMSC-EVs) naturally enriched with activated microglia-modulating miRNAs can avert chronic adverse outcomes of TBI. Small RNA sequencing confirmed the enrichment of miRNAs capable of modulating activated microglia in hMSC-EV cargo. IN administration of hMSC-EVs into adult mice ninety minutes after the induction of a unilateral controlled cortical impact injury resulted in their incorporation into neurons and microglia in both injured and contralateral hemispheres. A single higher dose hMSC-EV treatment also inhibited NLRP3 inflammasome activation after TBI, evidenced by reduced NLRP3, apoptosis-associated speck-like protein containing a CARD, activated caspase-1, interleukin-1 beta, and IL-18 levels in the injured brain. Such inhibition in the acute phase of TBI endured in the chronic phase, which could also be gleaned from diminished NLRP3 inflammasome activation in microglia of TBI mice receiving hMSC-EVs. Proteomic analysis and validation revealed that higher dose hMSC-EV treatment thwarted the chronic activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway by IL-18, which decreased the release of proinflammatory cytokines. Inhibition of the chronic activation of NLRP3-p38/MAPK signaling after TBI also prevented long-term cognitive and mood impairments. Notably, the animals receiving higher doses of hMSC-EVs after TBI displayed better cognitive and mood function in all behavioral tests than animals receiving the vehicle after TBI. A lower dose of hMSC-EV treatment also partially improved cognitive and mood function. Thus, an optimal IN dose of hMSC-EVs naturally enriched with activated microglia-modulating miRNAs can inhibit the chronic activation of NLRP3-p38/MAPK signaling after TBI and prevent lasting brain dysfunction.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Roxanne L Reger
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Bojana Milutinovic
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Jenny J Gonzalez
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Daniel L G Gitai
- Institute of Biological Sciences and Health, Federal University of Alagoas, Brazil
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Jong M Choi
- Advanced Technology Core, Mass Spectrometry and Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Sung Y Jung
- The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
| |
Collapse
|
16
|
Haltom AR, Hassen WE, Hensel J, Kim J, Sugimoto H, Li B, McAndrews KM, Conner MR, Kirtley ML, Luo X, Xie B, Volpert OV, Olalekan S, Maltsev N, Basu A, LeBleu VS, Kalluri R. Engineered exosomes targeting MYC reverse the proneural-mesenchymal transition and extend survival of glioblastoma. EXTRACELLULAR VESICLE 2022; 1:100014. [PMID: 37503329 PMCID: PMC10373511 DOI: 10.1016/j.vesic.2022.100014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dysregulated Myc signaling is a key oncogenic pathway in glioblastoma multiforme (GBM). Yet, effective therapeutic targeting of Myc continues to be challenging. Here, we demonstrate that exosomes generated from human bone marrow mesenchymal stem cells (MSCs) engineered to encapsulate siRNAs targeting Myc (iExo-Myc) localize to orthotopic GBM tumors in mice. Treatment of late stage GBM tumors with iExo-Myc inhibits proliferation and angiogenesis, suppresses tumor growth, and extends survival. Transcriptional profiling of tumors reveals that the mesenchymal transition and estrogen receptor signaling pathways are impacted by Myc inhibition. Single nuclei RNA sequencing (snRNA-seq) shows that iExo-Myc treatment induces transcriptional repression of multiple growth factor and interleukin signaling pathways, triggering a mesenchymal to proneural transition and shifting the cellular landscape of the tumor. These data confirm that Myc is an effective anti-glioma target and that iExo-Myc offers a feasible, readily translational strategy to inhibit challenging oncogene targets for the treatment of brain tumors.
Collapse
Affiliation(s)
- Amanda R. Haltom
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wafa E. Hassen
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Janine Hensel
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jiha Kim
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hikaru Sugimoto
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bingrui Li
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Meagan R. Conner
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michelle L. Kirtley
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Bioengineering, Rice University, Houston, TX
| | - Bingqing Xie
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Olga V. Volpert
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Susan Olalekan
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Natalia Maltsev
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Anindita Basu
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Valerie S. LeBleu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
- Feinberg School of Medicine & Kellogg School of Management, Northwestern University, Chicago, IL
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
- James P. Allison Institute at MD Anderson, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Department of Bioengineering, Rice University, Houston, TX
| |
Collapse
|
17
|
Baratta AM, Mangieri RA, Aziz HC, Lopez MF, Farris SP, Homanics GE. Effect of chronic intermittent ethanol vapor exposure on RNA content of brain-derived extracellular vesicles. Alcohol 2022; 105:9-24. [PMID: 36055466 PMCID: PMC10173183 DOI: 10.1016/j.alcohol.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 01/26/2023]
Abstract
Extracellular vesicles (EVs) are important players in normal biological function and disease pathogenesis. Of the many biomolecules packaged into EVs, coding and noncoding RNA transcripts are of particular interest for their ability to significantly alter cellular and molecular processes. Here we investigate how chronic ethanol exposure impacts EV RNA cargo and the functional outcomes of these changes. Following chronic intermittent ethanol (CIE) vapor exposure, EVs were isolated from male and female C57BL/6J mouse brain. Total RNA from EVs was analyzed by lncRNA/mRNA microarray to survey changes in RNA cargo following vapor exposure. Differential expression analysis of microarray data revealed a number of lncRNA and mRNA types differentially expressed in CIE compared to control EVs. Weighted gene co-expression network analysis identified multiple male and female specific modules related to neuroinflammation, cell death, demyelination, and synapse organization. To functionally test these changes, whole-cell voltage-clamp recordings were used to assess synaptic transmission. Incubation of nucleus accumbens brain slices with EVs led to a reduction in spontaneous excitatory postsynaptic current amplitude, although no changes in synaptic transmission were observed between control and CIE EV administration. These results indicate that CIE vapor exposure significantly changes the RNA cargo of brain-derived EVs, which have the ability to impact neuronal function.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Regina A Mangieri
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Heather C Aziz
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Science, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Sean P Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Gregg E Homanics
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
18
|
Sun K, Zheng X, Jin H, Yu F, Zhao W. Exosomes as CNS Drug Delivery Tools and Their Applications. Pharmaceutics 2022; 14:pharmaceutics14102252. [PMID: 36297688 PMCID: PMC9609403 DOI: 10.3390/pharmaceutics14102252] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Central nervous system (CNS) diseases threaten the health of people all over the world. However, due to the structural and functional particularities of the brain and spinal cord, CNS-targeted drug development is rather challenging. Exosomes are small cellular vesicles with lipid bilayers that can be secreted by almost all cells and play important roles in intercellular communication. The advantages of low immunogenicity, the ability to cross the blood-brain barrier, and the flexibility of drug encapsulation make them stand out among CNS drug delivery tools. Herein, we reviewed the research on exosomes in CNS drug delivery over the past decade and outlined the impact of the drug loading mode, administration route, and engineered modification on CNS targeting. Finally, we highlighted the problems and prospects of exosomes as CNS drug delivery tools.
Collapse
Affiliation(s)
- Ke Sun
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Xue Zheng
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Hongzhen Jin
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
- Correspondence: (H.J.); (F.Y.)
| | - Fan Yu
- College of Life Sciences, Nankai University, Weijin Road, Nankai District, Tianjin 300350, China
- Correspondence: (H.J.); (F.Y.)
| | - Wei Zhao
- College of Pharmacy, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| |
Collapse
|
19
|
Yang YS, He SL, Chen WC, Wang CM, Huang QM, Shi YC, Lin S, He HF. Recent progress on the role of non-coding RNA in postoperative cognitive dysfunction. Front Cell Neurosci 2022; 16:1024475. [PMID: 36313620 PMCID: PMC9608859 DOI: 10.3389/fncel.2022.1024475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD), especially in elderly patients, is a serious complication characterized by impairment of cognitive and sensory modalities after surgery. The pathogenesis of POCD mainly includes neuroinflammation, neuronal apoptosis, oxidative stress, accumulation of Aβ, and tau hyperphosphorylation; however, the exact mechanism remains unclear. Non-coding RNA (ncRNA) may play an important role in POCD. Some evidence suggests that microRNA, long ncRNA, and circular RNA can regulate POCD-related processes, making them promising biomarkers in POCD diagnosis, treatment, and prognosis. This article reviews the crosstalk between ncRNAs and POCD, and systematically discusses the role of ncRNAs in the pathogenesis and diagnosis of POCD. Additionally, we explored the possible mechanisms of ncRNA-associated POCD, providing new knowledge for developing ncRNA-based treatments for POCD.
Collapse
Affiliation(s)
- Yu-Shen Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shi-Ling He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiao-Mei Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-Chuan Shi
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Yan-Chuan Shi,
| | - Shu Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Shu Lin,
| | - He-fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- He-fan He,
| |
Collapse
|
20
|
Li C, Hu J, Liu W, Ke C, Huang C, Bai Y, Pan B, Wang J, Wan C. Exercise Intervention Modulates Synaptic Plasticity by Inhibiting Excessive Microglial Activation via Exosomes. Front Cell Neurosci 2022; 16:953640. [PMID: 35928570 PMCID: PMC9345504 DOI: 10.3389/fncel.2022.953640] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background Exosomes can activate microglia to modulate neural activity and synaptic plasticity by phagocytosis of neural spines or synapses. Our previous research found that an early 4-week exercise intervention in middle cerebral artery occlusion (MCAO) rats can promote the release of exosomes and protect the brain. This study intended to further explore the intrinsic mechanism of neuroprotection by exosome release after exercise. Methods Rats were randomly divided into four groups: the sham operation (SHAM), middle cerebral artery occlusion (MCAO) with sedentary intervention (SED-MCAO), MCAO with exercise intervention (EX-MCAO), and MCAO with exercise intervention and exosome injection (EX-MCAO-EXO). Modified neurological severity score (mNSS), cerebral infarction volume ratio, microglial activation, dendritic complexity, and expression of synaptophysin (Syn) and postsynaptic density protein 95 (PSD-95) were detected after 28 days of intervention. Results (1) The exercise improved body weight and mNSS score, and the survival state of the rats after exosome infusion was better. (2) Compared with the SED-MCAO group, the EX-MCAO (P = 0.039) and EX-MCAO-EXO groups (P = 0.002) had significantly lower cerebral infarct volume ratios (P < 0.05), among which the EX-MCAO-EXO group had the lowest (P = 0.031). (3) Compared with the SED-MCAO group, the EX-MCAO and EX-MCAO-EXO groups had a significantly decreased number of microglia (P < 0.001) and significantly increased process length/cell (P < 0.01) and end point/cell (P < 0.01) values, with the EX-MCAO-EXO group having the lowest number of microglia (P = 0.036) and most significantly increased end point/cell value (P = 0.027). (4) Compared with the SED-MCAO group, the total number of intersections and branches of the apical and basal dendrites in the EX-MCAO and EX-MCAO-EXO groups was increased significantly (P < 0.05), and the increase was more significant in the EX-MCAO-EXO group (P < 0.05). (5) The expression levels of Syn and PSD-95 in the EX-MCAO (PSyn = 0.043, PPSD−95 = 0.047) and EX-MCAO-EXO groups were significantly higher than those in the SED-MCAO group (P < 0.05), and the expression levels in the EX-MCAO-EXO group were significantly higher than those in the EX-MCAO group (P < 0.05). Conclusion Early exercise intervention after stroke can inhibit the excessive activation of microglia and regulate synaptic plasticity by exosome release.
Collapse
Affiliation(s)
- Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayi Hu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wenhong Liu
- Tianjin Rehabilitation Center, Tianjin, China
| | - Changkai Ke
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuan Huang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yifan Bai
- Department of Rehabilitation Medicine, School of Medicine Technology, Tianjin Medical University, Tianjin, China
| | - Bingchen Pan
- Department of Rehabilitation Medicine, School of Medicine Technology, Tianjin Medical University, Tianjin, China
| | - Junyi Wang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Chunxiao Wan
| |
Collapse
|
21
|
Rezabakhsh A, Mahdipour M, Nourazarian A, Habibollahi P, Sokullu E, Avci ÇB, Rahbarghazi R. Application of exosomes for the alleviation of COVID-19-related pathologies. Cell Biochem Funct 2022; 40:430-438. [PMID: 35647674 PMCID: PMC9348296 DOI: 10.1002/cbf.3720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 12/17/2022]
Abstract
The pandemic of COVID-19 caused worldwide concern. Due to the lack of appropriate medications and the inefficiency of commercially available vaccines, lots of efforts are being made to develop de novo therapeutic modalities. Besides this, the possibility of several genetic mutations in the viral genome has led to the generation of resistant strains such as Omicron against neutralizing antibodies and vaccines, leading to worsening public health status. Exosomes (Exo), nanosized vesicles, possess several therapeutic properties that participate in intercellular communication. The discovery and application of Exo in regenerative medicine have paved the way for the alleviation of several pathologies. These nanosized particles act as natural bioshuttles and transfer several biomolecules and anti-inflammatory cytokines. To date, several approaches are available for the administration of Exo into the targeted site inside the body, although the establishment of standard administration routes remains unclear. As severe acute respiratory syndrome coronavirus 2 primarily affects the respiratory system, we here tried to highlight the transplantation of Exo in the alleviation of COVID-19 pathologies.
Collapse
Affiliation(s)
- Aysa Rezabakhsh
- Cardiovascular Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mahdi Mahdipour
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
| | - Alireza Nourazarian
- Department of Basic Medical SciencesKhoy University of Medical SciencesKhoyIran
| | - Paria Habibollahi
- Department of Pharmacology and Toxicology, Faculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Emel Sokullu
- Koç University Research Center for Translational Medicine (KUTTAM)IstanbulSariyerTurkey
| | - Çigir Biray Avci
- Department of Medical Biology, Faculty of MedicineEge UniversityIzmirTurkey
| | - Reza Rahbarghazi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
22
|
Sharif NA. Degeneration of retina-brain components and connections in glaucoma: Disease causation and treatment options for eyesight preservation. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100037. [PMID: 36685768 PMCID: PMC9846481 DOI: 10.1016/j.crneur.2022.100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/25/2023] Open
Abstract
Eyesight is the most important of our sensory systems for optimal daily activities and overall survival. Patients who experience visual impairment due to elevated intraocular pressure (IOP) are often those afflicted with primary open-angle glaucoma (POAG) which slowly robs them of their vision unless treatment is administered soon after diagnosis. The hallmark features of POAG and other forms of glaucoma are damaged optic nerve, retinal ganglion cell (RGC) loss and atrophied RGC axons connecting to various brain regions associated with receipt of visual input from the eyes and eventual decoding and perception of images in the visual cortex. Even though increased IOP is the major risk factor for POAG, the disease is caused by many injurious chemicals and events that progress slowly within all components of the eye-brain visual axis. Lowering of IOP mitigates the damage to some extent with existing drugs, surgical and device implantation therapeutic interventions. However, since multifactorial degenerative processes occur during aging and with glaucomatous optic neuropathy, different forms of neuroprotective, nutraceutical and electroceutical regenerative and revitalizing agents and processes are being considered to combat these eye-brain disorders. These aspects form the basis of this short review article.
Collapse
Affiliation(s)
- Najam A. Sharif
- Duke-National University of Singapore Medical School, Singapore,Singapore Eye Research Institute (SERI), Singapore,Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA,Department of Pharmaceutical Sciences, Texas Southern University, Houston, TX, USA,Department of Surgery & Cancer, Imperial College of Science and Technology, St. Mary's Campus, London, UK,Department of Pharmacy Sciences, School of School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA.
| |
Collapse
|
23
|
Cell-Derived Exosomes as Therapeutic Strategies and Exosome-Derived microRNAs as Biomarkers for Traumatic Brain Injury. J Clin Med 2022; 11:jcm11113223. [PMID: 35683610 PMCID: PMC9181755 DOI: 10.3390/jcm11113223] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a complex, life-threatening condition that causes mortality and disability worldwide. No effective treatment has been clinically verified to date. Achieving effective drug delivery across the blood–brain barrier (BBB) presents a major challenge to therapeutic drug development for TBI. Furthermore, the field of TBI biomarkers is rapidly developing to cope with the many aspects of TBI pathology and enhance clinical management of TBI. Exosomes (Exos) are endogenous extracellular vesicles (EVs) containing various biological materials, including lipids, proteins, microRNAs, and other nucleic acids. Compelling evidence exists that Exos, such as stem cell-derived Exos and even neuron or glial cell-derived Exos, are promising TBI treatment strategies because they pass through the BBB and have the potential to deliver molecules to target lesions. Meanwhile, Exos have decreased safety risks from intravenous injection or orthotopic transplantation of viable cells, such as microvascular occlusion or imbalanced growth of transplanted cells. These unique characteristics also create Exos contents, especially Exos-derived microRNAs, as appealing biomarkers in TBI. In this review, we explore the potential impact of cell-derived Exos and exosome-derived microRNAs on the diagnosis, therapy, and prognosis prediction of TBI. The associated challenges and opportunities are also discussed.
Collapse
|
24
|
Patel RS, Impreso S, Lui A, Vidyarthi G, Albear P, Patel NA. Long Noncoding RNA GAS5 Contained in Exosomes Derived from Human Adipose Stem Cells Promotes Repair and Modulates Inflammation in a Chronic Dermal Wound Healing Model. BIOLOGY 2022; 11:biology11030426. [PMID: 35336800 PMCID: PMC8945809 DOI: 10.3390/biology11030426] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 01/17/2023]
Abstract
Simple Summary Wounds due to cuts, lacerations, or surgical incisions undergo healing through a highly regulated process. Occasionally, the skin is unable to heal in a timely manner, leading to chronic wounds and related sequelae, such as scarring, risk of infections on open wounds, and—as a growing body of evidence attests—psychological impact on the individual. In addition, certain diseases, such as diabetes, obesity, and cancer, are characterized by an ongoing state of very low-grade inflammation. This underlying inflammation substantially hinders wound healing. To improve the outcome of chronic wounds, we harvested the potential of exosomes (nanovesicles) secreted from human adipose stem cells. We demonstrate that exosomes are efficiently taken up by skin cells and promote healing by significantly accelerating wound closure time. To understand the mechanism by which exosomes promote wound healing, we identified an RNA called GAS5 that is a driver of the regenerative properties of exosomes. Additionally, we identified the inflammation pathways that are regulated by GAS5 to promote the healing of wounds. Such a determination is essential to move exosome therapy into the clinic. In conclusion, our results demonstrate that exosomes harvested from human adipose stem cells accelerate the healing of chronic recalcitrant wounds and thus have a tremendous therapeutic potential in wound healing. Abstract Chronic recalcitrant wounds result from delayed or slowed healing processes. Underlying inflammation is a substantial risk factor for impaired dermal wound healing and often leads to chronic wound-related sequelae. Human adipose stem cells (hASCs) have shown tremendous potential in regenerative medicine. The goal of this project was to improve the outcome of chronic wounds by harvesting the exosomes from hASCs for therapeutic intervention. The results demonstrate that long noncoding RNA GAS5 is highly enriched in hASC exosomes and, further, that GAS5 is central to promoting wound repair in vitro. To evaluate the outcome of wound healing in a chronic low-grade inflammatory environment, lipopolysaccharide-treated HDF cells were evaluated for their response to hASC exosome treatment. Ingenuity pathway analysis identified inflammation pathways and genes affected by exosomes in a GAS5-dependent manner. Using siRNA to deplete GAS5 in HDF, the results demonstrated that Toll-like receptor 7 (TLR7) expression levels were regulated by GAS5. Importantly, the results demonstrate that GAS5 regulates inflammatory pathway genes in a chronic inflammation environment. The results presented here demonstrate that hASC exosomes are a viable therapeutic that accelerate the healing of chronic recalcitrant wounds.
Collapse
Affiliation(s)
- Rekha S. Patel
- James A. Haley Veteran’s Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (R.S.P.); (S.I.); (G.V.); (P.A.)
| | - Sabrina Impreso
- James A. Haley Veteran’s Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (R.S.P.); (S.I.); (G.V.); (P.A.)
| | - Ashley Lui
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Gitanjali Vidyarthi
- James A. Haley Veteran’s Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (R.S.P.); (S.I.); (G.V.); (P.A.)
| | - Paul Albear
- James A. Haley Veteran’s Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (R.S.P.); (S.I.); (G.V.); (P.A.)
| | - Niketa A. Patel
- James A. Haley Veteran’s Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (R.S.P.); (S.I.); (G.V.); (P.A.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
- Correspondence: or ; Tel.: +1-813-972-2000 (ext. 7283)
| |
Collapse
|
25
|
Li N, Wang R, Ai X, Guo X, Liu J, Sun L, Zhang R. Effect of acupuncture treatment on cognitive impairment after traumatic brain injury in adults: A protocol for systematic review. Medicine (Baltimore) 2021; 100:e28451. [PMID: 34941200 PMCID: PMC8702273 DOI: 10.1097/md.0000000000028451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Acupuncture has been widely used to treat cognitive impairment after traumatic brain injury (TBI). But its efficiency has not been scientifically and methodically evaluated. The objective of this study is to evaluate the efficiency and safety of the acupuncture treatment for cognitive impairment after TBI in adults. METHODS This protocol of systematic review will be conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-analysis Protocols. We will conduct the literature searching in the following electronic databases: the Cochrane Library, MEDLINE, EMBASE, Web of Science, Springer, the Chinese Science Citation Database (CSCD), China National Knowledge Infrastructure (CNKI), the Chinese Biomedical Literature Database (CBM), Wanfang, and the Chinese Scientific Journal Database (VIP). The time limit for retrieving studies is from establishment to November 2021 for each database. All published randomized controlled trials related to this review will be included. Review Manager (V.5.3.5) will be implemented for the assessment of bias risk and data analyses. The selection of the studies, data abstraction, and validations will be performed independently by 2 researchers. RESULTS This review will assess the clinical efficacy and safety, as well as the acupoints characteristics of acupuncture on CI of TBI in adults. CONCLUSION This review will summarize the current evidence of acupuncture on CI of TBI outcomes and provide guidance for clinicians and patients to select acupuncture for CI of TBI in adults. TRAIL REGISTRATION NUMBER This protocol of systematic review has been registered on INPLASY website (No. INPLASY2021110113).
Collapse
Affiliation(s)
- Na Li
- School of Acupuncture-Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Ruihui Wang
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Xia Ai
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Xinrong Guo
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Juan Liu
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Lei Sun
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Rongchao Zhang
- School of Acupuncture-Tuina, Shaanxi University of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| |
Collapse
|
26
|
Rohden F, Teixeira LV, Bernardi LP, Ferreira PCL, Colombo M, Teixeira GR, de Oliveira FDS, Cirne Lima EO, Guma FCR, Souza DO. Functional Recovery Caused by Human Adipose Tissue Mesenchymal Stem Cell-Derived Extracellular Vesicles Administered 24 h after Stroke in Rats. Int J Mol Sci 2021; 22:12860. [PMID: 34884665 PMCID: PMC8657917 DOI: 10.3390/ijms222312860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability, intensely demanding innovative and accessible therapeutic strategies. Approaches presenting a prolonged period for therapeutic intervention and new treatment administration routes are promising tools for stroke treatment. Here, we evaluated the potential neuroprotective properties of nasally administered human adipose tissue mesenchymal stem cell (hAT-MSC)-derived extracellular vesicles (EVs) obtained from healthy individuals who underwent liposuction. After a single intranasal EV (200 µg/kg) administered 24 h after a focal permanent ischemic stroke in rats, a higher number of EVs, improvement of the blood-brain barrier, and re-stabilization of vascularization were observed in the recoverable peri-infarct zone, as well as a significant decrease in infarct volume. In addition, EV treatment recovered long-term motor (front paws symmetry) and behavioral impairment (short- and long-term memory and anxiety-like behavior) induced by ischemic stroke. In line with these findings, our work highlights hAT-MSC-derived EVs as a promising therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Francieli Rohden
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luciele Varaschini Teixeira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luis Pedro Bernardi
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Faculty of Biomedicine, Universidade Federal de Ciências da Saúde de Porto Alegre—UFCSPA, Porto Alegre 90050-170, Brazil
| | - Pamela Cristina Lukasewicz Ferreira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Mariana Colombo
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil;
| | - Geciele Rodrigues Teixeira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fernanda dos Santos de Oliveira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Elizabeth Obino Cirne Lima
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fátima Costa Rodrigues Guma
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Diogo Onofre Souza
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| |
Collapse
|